Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Virol ; 97(3): e0000323, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36877049

RESUMO

Pseudorabies virus (PRV) infection activates inflammatory responses to release robust proinflammatory cytokines, which are critical for controlling viral infection and clearance of PRV. However, the innate sensors and inflammasomes involved in the production and secretion of proinflammatory cytokines during PRV infection remain poorly studied. In this study, we report that the transcription and expression levels of some proinflammatory cytokines, including interleukin 1ß (IL-1ß), IL-6, and tumor necrosis factor alpha (TNF-α), are upregulated in primary peritoneal macrophages and in mice during PRV infection. Mechanistically, Toll-like receptor 2 (TLR2), TLR3, TLR4, and TLR5 were induced by the PRV infection to enhance the transcription levels of pro-IL-1ß, pro-IL-18, and gasdermin D (GSDMD). Additionally, we found that PRV infection and transfection of its genomic DNA triggered AIM2 inflammasome activation, apoptosis-related speckle-like protein (ASC) oligomerization, and caspase-1 activation to enhance the secretion of IL-1ß and IL-18, which was mainly dependent on GSDMD, but not GSDME, in vitro and in vivo. Taken together, our findings reveal that the activation of the TLR2-TLR3-TRL4-TLR5-NF-κB axis and AIM2 inflammasome, as well as GSDMD, is required for proinflammatory cytokine release, which resists the PRV replication and plays a critical role in host defense against PRV infection. Our findings provide novel clues to prevent and control PRV infection. IMPORTANCE PRV can infect several mammals, including pigs, other livestock, rodents, and wild animals, causing huge economic losses. As an emerging and reemerging infectious disease, the emergence of PRV virulent isolates and increasing human PRV infection cases indicate that PRV is still a high risk to public health. It has been reported that PRV infection leads to robust release of proinflammatory cytokines through activating inflammatory responses. However, the innate sensor that activates IL-1ß expression and the inflammasome involved in the maturation and secretion of proinflammatory cytokines during PRV infection remain poorly studied. In this study, our findings reveal that, in mice, activation of the TLR2-TLR3-TRL4-TLR5-NF-κB axis and AIM2 inflammasome, as well as GSDMD, is required for proinflammatory cytokine release during PRV infection, and it resists PRV replication and plays a critical role in host defense against PRV infection. Our findings provide novel clues to prevent and control PRV infection.


Assuntos
Herpesvirus Suídeo 1 , Inflamassomos , NF-kappa B , Animais , Humanos , Camundongos , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Herpesvirus Suídeo 1/metabolismo , Inflamassomos/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Mamíferos , NF-kappa B/metabolismo , Suínos , Receptor 2 Toll-Like/genética , Receptor 3 Toll-Like , Receptor 5 Toll-Like , Transdução de Sinais , Encefalite Viral/metabolismo
2.
Front Immunol ; 12: 753683, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899705

RESUMO

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Assuntos
Efeito Citopatogênico Viral , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Parechovirus/patogenicidade , Infecções por Picornaviridae/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Autofagia , Linhagem Celular Tumoral , Córtex Cerebral/virologia , Chlorocebus aethiops , Citocinas/biossíntese , Citocinas/genética , Encefalite Viral/patologia , Encefalite Viral/virologia , Glioblastoma/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/farmacologia , Interferons/biossíntese , Interferons/genética , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Parechovirus/efeitos dos fármacos , Parechovirus/fisiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Piroptose , Células Vero , Replicação Viral/efeitos dos fármacos , Interferon lambda
3.
Front Immunol ; 12: 667478, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025669

RESUMO

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Células Apresentadoras de Antígenos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encefalite Viral/prevenção & controle , Receptor Celular 2 do Vírus da Hepatite A/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Infecções por Rhabdoviridae/prevenção & controle , Vesiculovirus/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/virologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Chlorocebus aethiops , Modelos Animais de Doenças , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Encefalite Viral/virologia , Células HEK293 , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Injeções Intraperitoneais , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Células RAW 264.7 , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/metabolismo , Infecções por Rhabdoviridae/virologia , Ubiquitinação , Células Vero , Vesiculovirus/patogenicidade , Carga Viral
4.
Neuromolecular Med ; 23(1): 25-46, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33547562

RESUMO

Neuro-inflammation accompanies numerous neurological disorders and conditions where it can be associated with a progressive neurodegenerative pathology. In a similar manner, alterations in sphingolipid metabolism often accompany or are causative features in degenerative neurological conditions. These include dementias, motor disorders, autoimmune conditions, inherited metabolic disorders, viral infection, traumatic brain and spinal cord injury, psychiatric conditions, and more. Sphingolipids are major regulators of cellular fate and function in addition to being important structural components of membranes. Their metabolism and signaling pathways can also be regulated by inflammatory mediators. Therefore, as certain sphingolipids exert distinct and opposing cellular roles, alterations in their metabolism can have major consequences. Recently, regulation of bioactive sphingolipids by neuro-inflammatory mediators has been shown to activate a neuronal NADPH oxidase 2 (NOX2) that can provoke damaging oxidation. Therefore, the sphingolipid-regulated neuronal NOX2 serves as a mechanistic link between neuro-inflammation and neurodegeneration. Moreover, therapeutics directed at sphingolipid metabolism or the sphingolipid-regulated NOX2 have the potential to alleviate neurodegeneration arising out of neuro-inflammation.


Assuntos
NADPH Oxidase 2/metabolismo , Doenças Neurodegenerativas/metabolismo , Esfingolipídeos/fisiologia , Complexo AIDS Demência/metabolismo , Animais , Produtos Biológicos/uso terapêutico , Encefalopatias Metabólicas Congênitas/genética , Encefalopatias Metabólicas Congênitas/metabolismo , Encefalopatias Metabólicas Congênitas/terapia , Descoberta de Drogas , Encefalite Viral/metabolismo , Ativação Enzimática , Terapia de Reposição de Enzimas , Humanos , Inflamação , Naftalenos/uso terapêutico , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/terapia , Neurônios/metabolismo , Oxirredução , Pirimidinonas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Infecção por Zika virus/metabolismo
5.
J Virol ; 93(8)2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30728259

RESUMO

A clinical isolate of measles virus (MeV) bearing a single amino acid alteration in the viral fusion protein (F; L454W) was previously identified in two patients with lethal sequelae of MeV central nervous system (CNS) infection. The mutation dysregulated the viral fusion machinery so that the mutated F protein mediated cell fusion in the absence of known MeV cellular receptors. While this virus could feasibly have arisen via intrahost evolution of the wild-type (wt) virus, it was recently shown that the same mutation emerged under the selective pressure of small-molecule antiviral treatment. Under these conditions, a potentially neuropathogenic variant emerged outside the CNS. While CNS adaptation of MeV was thought to generate viruses that are less fit for interhost spread, we show that two animal models can be readily infected with CNS-adapted MeV via the respiratory route. Despite bearing a fusion protein that is less stable at 37°C than the wt MeV F, this virus infects and replicates in cotton rat lung tissue more efficiently than the wt virus and is lethal in a suckling mouse model of MeV encephalitis even with a lower inoculum. Thus, either during lethal MeV CNS infection or during antiviral treatment in vitro, neuropathogenic MeV can emerge, can infect new hosts via the respiratory route, and is more pathogenic (at least in these animal models) than wt MeV.IMPORTANCE Measles virus (MeV) infection can be severe in immunocompromised individuals and lead to complications, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE) occur even in the face of an intact immune response. While they are relatively rare complications of MeV infection, MIBE and SSPE are lethal. This work addresses the hypothesis that despite a dysregulated viral fusion complex, central nervous system (CNS)-adapted measles virus can spread outside the CNS within an infected host.


Assuntos
Sistema Nervoso Central/virologia , Encefalite Viral , Corpos de Inclusão Viral , Pulmão/virologia , Vírus do Sarampo/fisiologia , Sarampo , Mutação de Sentido Incorreto , Proteínas Virais de Fusão , Replicação Viral , Substituição de Aminoácidos , Animais , Sistema Nervoso Central/metabolismo , Chlorocebus aethiops , Modelos Animais de Doenças , Encefalite Viral/genética , Encefalite Viral/metabolismo , Encefalite Viral/transmissão , Humanos , Corpos de Inclusão Viral/genética , Corpos de Inclusão Viral/metabolismo , Pulmão/metabolismo , Sarampo/metabolismo , Sarampo/transmissão , Camundongos , Camundongos Transgênicos , Sigmodontinae , Células Vero , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
6.
Neuron ; 99(1): 56-63.e3, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-30001512

RESUMO

Amyloid-ß peptide (Aß) fibrilization and deposition as ß-amyloid are hallmarks of Alzheimer's disease (AD) pathology. We recently reported Aß is an innate immune protein that protects against fungal and bacterial infections. Fibrilization pathways mediate Aß antimicrobial activities. Thus, infection can seed and dramatically accelerate ß-amyloid deposition. Here, we show Aß oligomers bind herpesvirus surface glycoproteins, accelerating ß-amyloid deposition and leading to protective viral entrapment activity in 5XFAD mouse and 3D human neural cell culture infection models against neurotropic herpes simplex virus 1 (HSV1) and human herpesvirus 6A and B. Herpesviridae are linked to AD, but it has been unclear how viruses may induce ß-amyloidosis in brain. These data support the notion that Aß might play a protective role in CNS innate immunity, and suggest an AD etiological mechanism in which herpesviridae infection may directly promote Aß amyloidosis.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Encéfalo/metabolismo , Encefalite Viral/metabolismo , Herpesviridae , Doença de Alzheimer/virologia , Amiloidose/virologia , Animais , Encéfalo/virologia , Células Cultivadas , Modelos Animais de Doenças , Encefalite por Herpes Simples/metabolismo , Encefalite por Herpes Simples/virologia , Encefalite Viral/virologia , Herpesvirus Humano 1 , Herpesvirus Humano 6 , Humanos , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Neurônios , Placa Amiloide/metabolismo , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/virologia
7.
Neuron ; 99(1): 64-82.e7, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29937276

RESUMO

Investigators have long suspected that pathogenic microbes might contribute to the onset and progression of Alzheimer's disease (AD) although definitive evidence has not been presented. Whether such findings represent a causal contribution, or reflect opportunistic passengers of neurodegeneration, is also difficult to resolve. We constructed multiscale networks of the late-onset AD-associated virome, integrating genomic, transcriptomic, proteomic, and histopathological data across four brain regions from human post-mortem tissue. We observed increased human herpesvirus 6A (HHV-6A) and human herpesvirus 7 (HHV-7) from subjects with AD compared with controls. These results were replicated in two additional, independent and geographically dispersed cohorts. We observed regulatory relationships linking viral abundance and modulators of APP metabolism, including induction of APBB2, APPBP2, BIN1, BACE1, CLU, PICALM, and PSEN1 by HHV-6A. This study elucidates networks linking molecular, clinical, and neuropathological features with viral activity and is consistent with viral activity constituting a general feature of AD.


Assuntos
Doença de Alzheimer/virologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/virologia , Encefalite Viral/virologia , Herpesvirus Humano 6 , Herpesvirus Humano 7 , Infecções por Roseolovirus/virologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Clusterina/genética , Estudos de Coortes , Encefalite Viral/genética , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Genômica , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/genética , Microbiota , Proteínas Monoméricas de Montagem de Clatrina/genética , Proteínas Nucleares/genética , Presenilina-1/genética , Proteômica , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/patologia , Proteínas Supressoras de Tumor/genética , Carga Viral
8.
Clin Infect Dis ; 64(7): 914-920, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28362944

RESUMO

BACKGROUND: The unprecedented reemergence of Zika virus (ZIKV) has startled the world with reports of increased microcephaly in Brazil. ZIKV can infect human neural progenitors and impair brain growth. However, direct evidence of ZIKV infection in human fetal brain tissues remains elusive. METHODS: Investigations were performed with brain cell preparations obtained from 9 donors. Virus infectivity was assessed by detection of virus antigen by flow cytometry together with various hematopoietic cell surface markers. Virus replication was determined by viral RNA quantification. Cytokine levels in supernatant obtained from virus-infected fetal brain cells were measured simultaneously in microbead-based immunoassays. RESULTS: We also show that ZIKV infection was particularly evident in hematopoietic cells with microglia, the brain-resident macrophage population being one of the main targets. Infection induces high levels of proinflammatory immune mediators such as interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), interleukin 1ß (IL-1ß), and monocyte chemotactic protein 1 (MCP-1). CONCLUSIONS: Our results highlight an important role for microglia and neuroinflammation during congenital ZIKV pathogenesis.


Assuntos
Microglia/virologia , Infecção por Zika virus/virologia , Zika virus , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Citocinas/metabolismo , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Encefalite Viral/virologia , Feto , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Microcefalia/etiologia , Microglia/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Células Vero , Carga Viral , Infecção por Zika virus/imunologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia
9.
Virus Res ; 227: 220-230, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27794455

RESUMO

OBJECTIVES: Human Immunodeficiency Virus (HIV) infection can induce neurocognitive complications classified as HIV-associated neurocognitive disorder (HAND). The chitinase family is associated with innate immunity cells and many infectious diseases. METHODS: We analyzed microarray datasets obtained from NCBI in order to verify the expression of chitinase family genes in hippocampus of uninfected rhesus macaques versus those with histopathologic evidence of Simian Immunodeficiency Virus Encephalitis (SIVE). Moreover, we have analysed two human microarray datasets to verify the results obtained in macaques hippocampus affected by SIVE. For these studies, we have also used the open source tools Genome-scale Integrated Analysis of gene Networks in Tissues (GIANT) to identify the chitinase genes network. RESULTS: CHIT1, CHI3L1 and CHI3L2 levels were significantly increased in SIVE hippocampus as compared to non-infected control specimens. Furthermore, we found a negative correlation between CHIA vs. Brain Viral Load (BVL). These data was confirmed partially in human brain section of HAD/HIVE subjects. Also, we showed that HIV-1 was able to modulate the expression of CHIT1, CHI3L1, CHI3L2 and CHID1 in human macrophages. CONCLUSIONS: These results suggest that chitinase gene expression is altered in SIVE and in HAD/HIVE brain sections and call for more studies examining whether this is a protective immunological reaction or a destructive tissue response to encephalitis.


Assuntos
Quitinases/genética , Encefalite Viral/genética , Encefalite Viral/virologia , Expressão Gênica , HIV-1 , Vírus da Imunodeficiência Símia , Animais , Disfunção Cognitiva , Suscetibilidade a Doenças , Encefalite Viral/metabolismo , Encefalite Viral/fisiopatologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Hipocampo/metabolismo , Hipocampo/virologia , Humanos , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Família Multigênica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Carga Viral
10.
J Neurovirol ; 22(6): 866-870, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27400929

RESUMO

We previously reported that mRNA expression of the unique alternatively spliced OPRM1 isoform µ-opioid receptor-1K (MOR-1K), which exhibits excitatory cellular signaling, is elevated in HIV-infected individuals with combined neurocognitive impairment (NCI) and HIV encephalitis (HIVE). It has recently been shown that the ß2-adrenergic receptor (ß2-AR) chaperones MOR-1K, normally localized intracellularly, to the cell surface. Here, we found mRNA expression of the adrenoceptor beta 2 (ADRB2) gene is also elevated in NCI-HIVE individuals, as well as that ß2-AR protein expression is elevated in HIV-1-infected primary human astrocytes treated with morphine, and discuss the implications for MOR-1K subcellular localization in this condition.


Assuntos
Complexo AIDS Demência/genética , Encefalite Viral/genética , RNA Mensageiro/genética , Receptores Adrenérgicos beta 2/genética , Receptores Opioides mu/genética , Complexo AIDS Demência/metabolismo , Complexo AIDS Demência/patologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Regulação da Expressão Gênica , Humanos , Morfina/farmacologia , Cultura Primária de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/metabolismo , Substância Branca/metabolismo , Substância Branca/patologia
11.
Neurobiol Dis ; 92(Pt B): 137-43, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26851503

RESUMO

A hallmark of persistent HIV-1 infection in the central nervous system is increased activation of mononuclear phagocytes and surrounding astrogliosis, conferring persistent HIV-induced inflammation. This inflammation is believed to result in neuronal dysfunction and the clinical manifestations of HIV-associated neurocognitive disorders (HAND). The Jak/STAT pathway is activated in macrophages/myeloid cells upon HIV-1 infection, modulating many pro-inflammatory pathways that result in HAND, thereby representing an attractive cellular target. Thus, the impact of ruxolitinib, a Janus Kinase (Jak) 1/2 inhibitor that is FDA approved for myelofibrosis and polycythemia vera, was assessed for its potential to inhibit HIV-1 replication in macrophages and HIV-induced activation in monocytes/macrophages in culture. In addition, a murine model of HIV encephalitis (HIVE) was used to assess the impact of ruxolitinib on histopathological features of HIVE, brain viral load, as well as its ability to penetrate the blood-brain-barrier (BBB). Ruxolitinib was found to inhibit HIV-1 replication in macrophages, HIV-induced activation of monocytes (CD14/CD16) and macrophages (HLA-DR, CCR5, and CD163) without apparent toxicity. In vivo, systemically administered ruxolitinib was detected in the brain during HIVE in SCID mice and markedly inhibited astrogliosis. Together, these data indicate that ruxolitinib reduces HIV-induced activation and infiltration of monocytes/macrophages in vitro, reduces the replication of HIV in vitro, penetrates the BBB when systemically administered in mice and reduces astrogliosis in the brains of mice with HIVE. These data suggest that ruxolitinib will be useful as a novel therapeutic to treat humans with HAND.


Assuntos
Antivirais/farmacologia , Encefalite Viral/tratamento farmacológico , Infecções por HIV/tratamento farmacológico , Inibidores de Janus Quinases/farmacologia , Pirazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Astrócitos/virologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Gliose/tratamento farmacológico , Gliose/metabolismo , Gliose/patologia , Gliose/virologia , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Masculino , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/virologia , Nitrilas , Pirimidinas , Tenofovir/farmacologia
12.
Proteomics Clin Appl ; 10(2): 126-35, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26033855

RESUMO

PURPOSE: To map the phosphoproteome and identify changes in the phosphorylation patterns in the HIV-infected and uninfected brain. EXPERIMENTAL DESIGN: Parietal cortex from individuals with and without HIV infection were lysed and trypsinized. The peptides were labeled with iTRAQ reagents, combined, phospho-enriched by titanium dioxide chromatography, and analyzed by LC-MS/MS with high resolution. RESULTS: Our phosphoproteomic workflow resulted in the identification of 112 phosphorylated proteins and 17 novel phosphorylation sites in all the samples that were analyzed. The phosphopeptide sequences were searched for kinase substrate motifs, which revealed potential kinases involved in important signaling pathways. The site-specific phosphopeptide quantification showed that peptides from neurofilament medium polypeptide, myelin basic protein, and 2'-3'-cyclic nucleotide-3' phosphodiesterase have relatively higher phosphorylation levels during HIV infection. CONCLUSIONS AND CLINICAL RELEVANCE: This study has enriched the global phosphoproteome knowledge of the human brain by detecting novel phosphorylation sites on neuronal proteins and identifying differentially phosphorylated brain proteins during HIV infection. Kinases that lead to unusual phosphorylations could be therapeutic targets for the treatment of HIV-associated neurocognitive disorders.


Assuntos
Encéfalo/metabolismo , Encefalite Viral/metabolismo , Infecções por HIV/metabolismo , Fosfoproteínas/análise , Proteômica , Encéfalo/virologia , Encefalite Viral/virologia , Humanos , Espectrometria de Massas , Fosfoproteínas/metabolismo , Fosforilação
13.
J Neurovirol ; 22(1): 8-13, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25994917

RESUMO

Rabies is an acute encephalomyelitis in humans and animals caused by rabies virus (RABV) infection. Because the neuropathological changes are very mild in rabies, it has been assumed that neuronal dysfunction likely explains the severe clinical disease. Recently, degenerative changes have been observed in neuronal processes (dendrites and axons) in experimental rabies. In vitro studies have shown evidence of oxidative stress that is caused by mitochondrial dysfunction. Recent work has shown that the RABV phosphoprotein (P) interacts with mitochondrial Complex I leading to overproduction of reactive oxygen species, which results in injury to axons. Amino acids at positions 139 to 172 of the P are critical in this process. Rabies vectors frequently show behavioral changes. Aggressive behavior with biting is important for transmission of the virus to new hosts at a time when virus is secreted in the saliva. Aggression is associated with low serotonergic activity in the brain. Charlton and coworkers performed studies in experimentally infected striped skunks with skunk rabies virus and observed aggressive behavioral responses. Heavy accumulation of RABV antigen was found in the midbrain raphe nuclei, indicating that impaired serotonin neurotransmission from the brainstem may account for the aggressive behavior. We now have an improved understanding of how RABV causes neuronal injury and how the infection results in behavioral changes that promote viral transmission to new hosts.


Assuntos
Agressão , Encefalite Viral/virologia , Interações Hospedeiro-Patógeno , Núcleos da Rafe do Mesencéfalo/virologia , Neurônios/virologia , Vírus da Raiva/patogenicidade , Raiva/virologia , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Comportamento Animal , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Encefalite Viral/metabolismo , Encefalite Viral/fisiopatologia , Encefalite Viral/transmissão , Mephitidae/virologia , Núcleos da Rafe do Mesencéfalo/patologia , Núcleos da Rafe do Mesencéfalo/fisiopatologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitocôndrias/virologia , Chaperonas Moleculares , Neurônios/metabolismo , Neurônios/patologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Raiva/metabolismo , Raiva/fisiopatologia , Raiva/transmissão , Vírus da Raiva/genética , Espécies Reativas de Oxigênio/metabolismo , Serotonina/metabolismo , Transmissão Sináptica , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
14.
J Neurol Sci ; 349(1-2): 190-5, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25626769

RESUMO

We examined serum levels of various cytokines, chemokines, growth factors, and adhesion molecules in patients with uncomplicated influenza (n=20) and influenza virus-associated encephalopathy (IE) (n=18) to understand the underlying mechanism of IE. We found that IL-1ß, IL-2, IL-5, IL-6, IL-7, IL-8, IL-10, IL-13, G-CSF, GM-CSF, TNF-α, TIMP-1, MMP-9, sE-selectin, and neutrophil elastase were elevated significantly in sera from patients with uncomplicated influenza and those with IE, compared with normal controls (n=20). Of note, neutrophil elastase, sE-selectin, IL-8, and IL-13 were elevated significantly in IE as compared with uncomplicated influenza. In the present study, for the first time, we found that serum levels of neutrophil elastase were increased in patients with IE compared with uncomplicated influenza, which suggested that cerebral endothelial damage in the development of IE was mediated by neutrophil elastase. The present study implied that anti-elastase agents are possibly an effective therapeutic protocol for IE, but this needs further elucidation.


Assuntos
Encefalite Viral/imunologia , Influenza Humana/imunologia , Elastase de Leucócito/sangue , Criança , Pré-Escolar , Citocinas/sangue , Selectina E/sangue , Encefalite Viral/sangue , Encefalite Viral/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Humanos , Lactente , Influenza Humana/sangue , Influenza Humana/metabolismo , Influenza Humana/virologia , Interleucina-10/sangue , Interleucina-13/sangue , Interleucina-1beta/sangue , Interleucina-2/sangue , Interleucina-5/sangue , Interleucina-6/sangue , Interleucina-7/sangue , Interleucina-8/sangue , Masculino , Inibidor Tecidual de Metaloproteinase-1/sangue , Fator de Necrose Tumoral alfa/sangue
15.
Inflammation ; 38(2): 728-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25081498

RESUMO

Interleukin-6 (IL-6), as one of pro-inflammatory cytokines, plays a key role in Enterovirus 71 (EV71) encephalitis. We investigated the association of IL-6-572C/G polymorphism and serum or cerebrospinal fluid (CSF) IL-6 level with EV71 encephalitis in patients with hand, foot, and mouth disease (HFMD). This study was carried out in 59 Chinese Han patients with EV71 encephalitis, 128 EV71-related HFMD without complications, and 232 controls. The IL-6-572C/G polymorphism was detected by polymerase chain reaction-restricted fragment length polymorphism gene analysis. Serum or CSF IL-6 levels were determined using a commercial enzyme-linked immunosorbent assay. The patients with EV71 encephalitis had a higher frequency of IL-6-572GG/GC genotype compared to the patients with EV71-related HFMD without encephalitis complications (40.7 vs. 15.6 %, odds ratio (OR)=3.70, 95 % confidence interval (CI)=1.83-7.50, p=0.001). Similarly, the frequency of IL-6-572 G allele among the patients with EV71 encephalitis was also higher than that of patients with EV71-related HFMD without encephalitis complications (23.7 vs. 8.6 %, OR=3.31, 95 % CI=1.80-6.08, p<0.001). Serum IL-6 levels in G carries (CG + GG) (195.1 ± 11.8 pg/ml) elevated significantly compared to CC homozygotes (167.7 ± 6.7 pg/ml) in EV71-infected patients (p<0.001), but no significant differences were observed in CSF IL-6 levels among different genotypes in patients with EV71 encephalitis. Furthermore, G carriers (GG + GC) (10.6 ±.29 mg/l) had significantly higher blood CRP levels compared to CC homozygotes (9.31 ± 1.93 mg/l) in patients with EV71 encephalitis (p=0.005). These findings suggested that IL-6-572 G allele was significantly associated with the susceptibility to EV71 encephalitis in Chinese Han patients, and IL-6-572 G allele might elevate the risk to EV71 encephalitis.


Assuntos
Povo Asiático/genética , Encefalite Viral/genética , Enterovirus Humano A , Infecções por Enterovirus/genética , Doença de Mão, Pé e Boca/genética , Interleucina-6/genética , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Pré-Escolar , Encefalite Viral/diagnóstico , Encefalite Viral/metabolismo , Infecções por Enterovirus/diagnóstico , Infecções por Enterovirus/metabolismo , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença/genética , Doença de Mão, Pé e Boca/diagnóstico , Doença de Mão, Pé e Boca/metabolismo , Humanos , Interleucina-6/sangue , Interleucina-6/líquido cefalorraquidiano , Masculino , Polimorfismo Genético/genética
16.
Clin Transplant ; 28(5): 540-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24579822

RESUMO

Human herpesvirus-6 (HHV-6) encephalopathy is a serious complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although reactivation of HHV-6 is often observed after allo-HSCT, encephalopathy only affects a few patients with HHV-6 reactivation. Human leukocyte antigen (HLA) class I is expressed by most somatic cells, and a relationship between some class I alleles and neurological diseases has been reported. The HHV-6 load at two, three, and four weeks after allo-HSCT was examined. HHV-6 encephalopathy was diagnosed from symptoms, results of cerebrospinal fluid examination, and magnetic resonance imaging findings. The relation between HHV-6 reactivation or encephalopathy and the HLA class I status of the recipients was investigated. In 130 patients, 147 allo-HSCT transplantation procedures were carried out. HHV-6 reactivation and encephalopathy occurred in 56 and nine procedures, respectively. HLA mismatch (p = 0.008) and unrelated donor (p = 0.001) were associated with HHV-6 reactivation, but not with HHV-6 encephalopathy. HHV-6 encephalopathy was more frequent in patients with HLA-B*40:06 (p = 0.027). In addition, HLA-A*26:01 and HLA-B*40:06 were found to be associated with each other (p = 0.089), while HLA-B*40:06 and HLA-C*08:01 showed a significant association (p < 0.001). The HLA class I alleles of recipients may be associated with the occurrence of HHV-6 encephalopathy after allo-HSCT.


Assuntos
Encefalite Viral/etiologia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Herpesvirus Humano 6/patogenicidade , Antígenos de Histocompatibilidade Classe I/metabolismo , Infecções por Roseolovirus/etiologia , DNA Viral/genética , Encefalite Viral/metabolismo , Feminino , Seguimentos , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Infecções por Roseolovirus/metabolismo , Transplante Homólogo , Ativação Viral
17.
Neuroimage ; 92: 83-9, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24513151

RESUMO

Adenosine A1 receptors (A1Rs) are implied in the modulation of neuroinflammation. Activation of cerebral A1Rs acts as a brake on the microglial response after traumatic brain injury and has neuroprotective properties in animal models of Parkinson's disease and multiple sclerosis. Neuroinflammatory processes in turn may affect the expression of A1Rs, but the available data is limited and inconsistent. Here, we applied an animal model of encephalitis to assess how neuroinflammation affects the expression of A1Rs. Two groups of animals were studied: Infected rats (n=7) were intranasally inoculated with herpes simplex virus-1 (HSV-1, 1 × 10(7) plaque forming units), sham-infected rats (n=6) received only phosphate-buffered saline. Six or seven days later, microPET scans (60 min with arterial blood sampling) were made using the tracer 8-dicyclopropyl-1-(11)C-methyl-3-propyl-xanthine ((11)C-MPDX). Tracer clearance from plasma and partition coefficient (K1/k2 estimated from a 2-tissue compartment model fit) were not significantly altered after virus infection. PET tracer distribution volume calculated from a Logan plot was significantly increased in the hippocampus (+37%) and medulla (+27%) of virus infected rats. Tracer binding potential (k3/k4 estimated from the model fit) was significantly increased in the cerebellum (+87%) and the medulla (+148%) which may indicate increased A1R expression. This was confirmed by immunohistochemical analysis showing a strong increase of A1R immunoreactivity in the cerebellum of HSV-1-infected rats. Both the quantitative PET data and immunohistochemical analysis indicate that A1Rs are upregulated in brain areas where active virus is present.


Assuntos
Encéfalo/metabolismo , Encefalite Viral/metabolismo , Infecções por HIV/metabolismo , Fatores Imunológicos/metabolismo , Tomografia por Emissão de Pósitrons/veterinária , Receptor A1 de Adenosina/metabolismo , Xantinas/farmacocinética , Animais , Animais não Endogâmicos , Encefalite Viral/diagnóstico por imagem , Infecções por HIV/diagnóstico por imagem , Taxa de Depuração Metabólica , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Wistar , Distribuição Tecidual , Regulação para Cima
18.
Am J Trop Med Hyg ; 89(5): 1013-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24080631

RESUMO

Rocio virus (ROCV) caused an outbreak of human encephalitis during the 1970s in Brazil and its immunopathogenesis remains poorly understood. CC-chemokine receptor 5 (CCR5) is a chemokine receptor that binds to macrophage inflammatory protein (MIP-1 α). Both molecules are associated with inflammatory cells migration during infections. In this study, we demonstrated the importance of the CCR5 and MIP-1 α, in the outcome of viral encephalitis of ROCV-infected mice. CCR5 and MIP-1 α knockout mice survived longer than wild-type (WT) ROCV-infected animals. In addition, knockout mice had reduced inflammation in the brain. Assessment of brain viral load showed mice virus detection five days post-infection in wild-type and CCR5-/- mice, while MIP-1 α-/- mice had lower viral loads seven days post-infection. Knockout mice required a higher lethal dose than wild-type mice as well. The CCR5/MIP-1 α axis may contribute to migration of infected cells to the brain and consequently affect the pathogenesis during ROCV infection.


Assuntos
Encéfalo/patologia , Quimiocina CCL3/genética , Encefalite Viral/metabolismo , Infecções por Flavivirus/metabolismo , Flavivirus/fisiologia , Receptores CCR5/genética , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Movimento Celular , Quimiocina CCL3/deficiência , Encefalite Viral/mortalidade , Encefalite Viral/patologia , Encefalite Viral/virologia , Infecções por Flavivirus/mortalidade , Infecções por Flavivirus/patologia , Infecções por Flavivirus/virologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Inflamação/metabolismo , Inflamação/mortalidade , Inflamação/patologia , Inflamação/virologia , Linfócitos/metabolismo , Linfócitos/patologia , Linfócitos/virologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Receptores CCR5/deficiência , Transdução de Sinais , Análise de Sobrevida , Carga Viral
19.
JAMA Neurol ; 70(10): 1305-10, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23939463

RESUMO

IMPORTANCE: Idiopathic narcolepsy with cataplexy is thought to be an autoimmune disorder targeting hypothalamic hypocretin neurons. Symptomatic narcolepsy with low hypocretin level has been described in Ma antibody­associated encephalitis; however, the mechanisms underlying such an association remain unknown. OBSERVATIONS: We described a 63-year-old man with clinical criteria for diencephalic encephalitis with sleepiness, cataplexy, hypocretin deficiency, and central hypothyroidism, together with brainstem encephalitis reflected by supranuclear ophtalmoparesis and rapid eye movement sleep behavior disorder with underlying abnormalities on brain magnetic resonance imaging. An autoimmune process was demonstrated by the detection of antibodies against Ma protein. Death occurred 4 months after disease onset without any tumor detected. Neuropathology, immunohistochemistry, and immunoreactivity results were compared with those obtained in idiopathic narcolepsy-cataplexy and with normal control brains. The principal findings revealed almost exclusive inflammation and tissue injury in the hypothalamus. The type of inflammatory reaction suggests cytotoxic CD8+ T lymphocytes being responsible for the induction of tissue injury. Inflammation was associated with complete loss of hypocretinergic neurons. Autoantibodies of the patient predominantly stained neurons in the hypothalamus and could be absorbed with Ma2. CONCLUSIONS AND RELEVANCE: The encephalitic process, responsible for narcolepsy-cataplexy and hypocretin deficiency, reflects a CD8+ inflammatory-mediated response against hypocretin neurons.


Assuntos
Encefalite Viral , Antígenos HIV/imunologia , Hipotálamo/metabolismo , Narcolepsia/complicações , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Antígenos CD/metabolismo , Aquaporina 4/metabolismo , Autoanticorpos/sangue , Autoanticorpos/líquido cefalorraquidiano , Encefalite Viral/complicações , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neuropeptídeos/metabolismo , Orexinas , Terceiro Ventrículo/patologia
20.
Acta Histochem ; 115(5): 407-11, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23305876

RESUMO

The ß-galactoside-binding lectin, galectin-3, is expressed in a variety of mammalian cells and tissues. It is involved in cell adhesion, activation, proliferation, apoptosis and cell migration. It also plays an important role in inflammation as a pro-inflammatory mediator. The involvement of galectin-3 in various inflammation models, including those of autoimmune disease, skin disease, and cancer, has been investigated extensively. Moreover, galectin-3 has been suggested to be a therapeutic target for various diseases. The present review deals with the expression of galectin-3 in central nervous system (CNS) tissues during normal development and in various models of inflammation. The available information indicates that galectin-3 is essential for normal brain development and plays diverse roles in CNS inflammation, combining pro-inflammatory roles with re-modeling capacity in damaged CNS tissues.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Encefalite/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Galectina 3/metabolismo , Animais , Proteínas Sanguíneas , Encéfalo/crescimento & desenvolvimento , Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Encefalite/patologia , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Encefalomielite Autoimune Experimental/patologia , Galectinas , Humanos , Camundongos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA